Next Article in Journal
Presence of the Neurotoxin BMAA in Aquatic Ecosystems: What Do We Really Know?
Previous Article in Journal
Zearalenone, an Estrogenic Mycotoxin, Is an Immunotoxic Compound
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Stimulation of Erythrocyte Cell Membrane Scrambling by Mushroom Tyrosinase

Department of Physiology, Eberhard-Karls-University of Tuebingen, Gmelinstr. 5, 72076 Tübingen, Germany
*
Author to whom correspondence should be addressed.
Toxins 2014, 6(3), 1096-1108; https://doi.org/10.3390/toxins6031096
Submission received: 5 February 2014 / Revised: 24 February 2014 / Accepted: 5 March 2014 / Published: 18 March 2014

Abstract

:
Background: Mushroom tyrosinase, a copper containing enzyme, modifies growth and survival of tumor cells. Mushroom tyrosinase may foster apoptosis, an effect in part due to interference with mitochondrial function. Erythrocytes lack mitochondria but are able to undergo apoptosis-like suicidal cell death or eryptosis, which is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine-exposure at the erythrocyte surface. Signaling involved in the triggering of eryptosis include increase of cytosolic Ca2+-activity ([Ca2+]i) and activation of sphingomyelinase with subsequent formation of ceramide. The present study explored, whether tyrosinase stimulates eryptosis. Methods: Cell volume has been estimated from forward scatter, phosphatidylserine-exposure from annexin V binding, [Ca2+]i from Fluo3-fluorescence, and ceramide abundance from binding of fluorescent antibodies in flow cytometry. Results: A 24 h exposure to mushroom tyrosinase (7 U/mL) was followed by a significant increase of [Ca2+]i, a significant increase of ceramide abundance, and a significant increase of annexin-V-binding. The annexin-V-binding following tyrosinase treatment was significantly blunted but not abrogated in the nominal absence of extracellular Ca2+. Tyrosinase did not significantly modify forward scatter. Conclusions: Tyrosinase triggers cell membrane scrambling, an effect, at least partially, due to entry of extracellular Ca2+ and ceramide formation.

1. Introduction

Mushroom tyrosinase has been suggested for the use in malignancy [1]. When applied with appropriate substrates, it may generate cytostatic products effective in vivo [2,3,4]. Tyrosinase may at least in part be effective by interference with mitochondrial function [2]. On the other hand, mushroom tyrosinase generates products leading to mutagenesis and carcinogenesis [5,6,7,8,9,10]. As a matter of fact, tyrosinase has been shown to trigger the suicidal death of nucleated cells or apoptosis [11].
Even though lacking mitochondria and nuclei, erythrocytes are still able to undergo apoptosis-like suicidal death or eryptosis [12]. Eryptosis may be elicited by increase of cytosolic Ca2+ concentration ([Ca2+]i) resulting at least partially from Ca2+ entry through Ca2+-permeable cation channels [12]. An increase of [Ca2+]i may shrink erythrocytes due to activation of Ca2+-sensitive K+ channels leading to K+ exit, hyperpolarization, Cl- exit and, thus, cellular loss of KCl and osmotically obliged water [13]. Increased [Ca2+]i further stimulates cell membrane scrambling with translocation of phosphatidylserine to the erythrocyte surface [12]. The Ca2+ sensitivity of cell membrane scrambling is increased by ceramide [12]. Signaling of eryptosis further includes caspases [14,15,16,17,18] and several kinases including AMP activated kinase AMPK [19], casein kinase 1α [20,21], cGMP-dependent protein kinase [22], Janus-activated kinase JAK3 [23], protein kinase C [24], p38 kinase [25], PAK2 kinase [26], as well as sorafenib [27] and sunifinib [28] sensitive kinases.
Eryptosis is stimulated by a wide variety of xenobiotics [12,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63] and excessive eryptosis is observed in several clinical disorders [12], such as diabetes [18,64,65], renal insufficiency [66], hemolytic uremic syndrome [67], sepsis [68], malaria [69], sickle cell disease [70], Wilson’s disease [71], iron deficiency [72], malignancy [73], phosphate depletion [74], and metabolic syndrome [48].
The present study explored, whether tyrosinase influences [Ca2+]i, cell volume and phosphatidylserine translocation to the erythrocyte surface. The observations disclose that exposure to tyrosinase stimulates erythrocyte cell membrane scrambling, an effect paralleled by and at least in part secondary to increase of [Ca2+]i.

2. Results and Discussion

The present study addressed the effect of tyrosinase on eryptosis. A hallmark of eryptosis is the breakdown of phosphatidylserine asymmetry of the erythrocyte cell membrane, which increases the phosphatidylserine abundance at the cell surface. Phosphatidylserine exposing erythrocytes were identified by annexin-V-binding in FACS analysis. As illustrated in Figure 1, a 24-h exposure to tyrosinase increased the percentage of annexin-V-binding erythrocytes, an effect reaching statistical significance at 5 U/mL tyrosinase activity.
A second hallmark of eryptosis is cell shrinkage. Accordingly, cell volume was estimated utilizing forward scatter, which was determined by flow cytometry. As shown in Figure 2, a 24-h exposure to tyrosinase tended to increase erythrocyte forward scatter, an effect, however, not reaching statistical significance. Further experiments were performed to elucidate whether tyrosinase abrogates the effect of the Ca2+ ionophore inomomycin (1 µM) on erythrocytes forward scatter. As a result, a 30-min exposure of erythrocytes was followed by a decrease of forward scatter from 518 ± 6 (n = 4) to 134 ± 5 (n = 4) in the absence of tyrosinase and from 557 ± 8 (n = 4) to 194 ± 16 (n = 4) in the presence of 7 U/mL tyrosinase. Thus, tyrosinase did not abrogate the shrinking effect of excessive Ca2+ entry.
Figure 1. Effect of tyrosinase on phosphatidylserine exposure. (A,B) Original histogram of annexin V binding of erythrocytes following exposure for 24 h to Ringer solution without (A) or with (B) 7 U/mL tyrosinase. M1 indicates the gating of annexin V binding cells (C) Arithmetic means ± SEM (n = 4) of erythrocyte annexin-V-binding following incubation for 24 h to Ringer solution without (white bar) or with (black bars) presence of tyrosinase (1–7 U/mL). ** (p < 0.01), *** (p < 0.001) indicate significant differences from the absence of tyrosinase (ANOVA).
Figure 1. Effect of tyrosinase on phosphatidylserine exposure. (A,B) Original histogram of annexin V binding of erythrocytes following exposure for 24 h to Ringer solution without (A) or with (B) 7 U/mL tyrosinase. M1 indicates the gating of annexin V binding cells (C) Arithmetic means ± SEM (n = 4) of erythrocyte annexin-V-binding following incubation for 24 h to Ringer solution without (white bar) or with (black bars) presence of tyrosinase (1–7 U/mL). ** (p < 0.01), *** (p < 0.001) indicate significant differences from the absence of tyrosinase (ANOVA).
Toxins 06 01096 g001
Figure 2. Effect of tyrosinase on erythrocyte forward scatter. (A) Original histogram of forward scatter of erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence of 7 U/mL tyrosinase; (B) arithmetic means ± SEM (n = 4) of the normalized erythrocyte forward scatter (FSC) following incubation for 24 h to Ringer solution without (white bar) or with (black bars) tyrosinase (1–7 U/mL).
Figure 2. Effect of tyrosinase on erythrocyte forward scatter. (A) Original histogram of forward scatter of erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence of 7 U/mL tyrosinase; (B) arithmetic means ± SEM (n = 4) of the normalized erythrocyte forward scatter (FSC) following incubation for 24 h to Ringer solution without (white bar) or with (black bars) tyrosinase (1–7 U/mL).
Toxins 06 01096 g002
Cell membrane scrambling is stimulated by increase of cytosolic Ca2+ activity ([Ca2+]i). Thus, [Ca2+]i was determined utilizing Fluo3 fluorescence. To this end, erythrocytes were loaded with Fluo3-AM and Fluo3 fluorescence determined in FACS analysis following prior incubation in Ringer solution without or with tyrosinase. As shown in Figure 3, a 24-h exposure of human erythrocytes to tyrosinase was followed by an increase of Fluo3 fluorescence, an effect reaching statistical significance at 5 U/mL of tyrosinase concentration.
Figure 3. Effect of mushroom tyrosinase on erythrocyte cytosolic Ca2+ concentration. (A) Original histogram of Fluo3 fluorescence in erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence of 7 U/mL tyrosinase; (B) arithmetic means ± SEM (n = 4) of the Fluo3 fluorescence (arbitrary units) in erythrocytes exposed for 24 h to Ringer solution without (white bar) or with (black bars) tyrosinase (1–7 U/mL). * (p < 0.05) indicates significant difference from the absence of tyrosinase (ANOVA).
Figure 3. Effect of mushroom tyrosinase on erythrocyte cytosolic Ca2+ concentration. (A) Original histogram of Fluo3 fluorescence in erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence of 7 U/mL tyrosinase; (B) arithmetic means ± SEM (n = 4) of the Fluo3 fluorescence (arbitrary units) in erythrocytes exposed for 24 h to Ringer solution without (white bar) or with (black bars) tyrosinase (1–7 U/mL). * (p < 0.05) indicates significant difference from the absence of tyrosinase (ANOVA).
Toxins 06 01096 g003
An additional series of experiments was performed testing whether extracellular Ca2+ entry was required for the effect of tyrosinase on cell membrane scrambling. Erythrocytes were exposed to 7 U/mL tyrosinase for 24 h, either in the presence of 1 mM Ca2+, or in the absence of Ca2+, and the presence of Ca2+ chelator EGTA (1 mM). As illustrated in Figure 4, the effect of tyrosinase on annexin-V-binding was significantly decreased in the nominal absence of Ca2+. However, even in the absence of extracellular Ca2+ tyrosinase still increased the percentage annexin V binding erythrocytes.
In order to test whether tyrosinase increased the formation of ceramide, which is known to trigger eryptosis even without increase of [Ca2+]i, ceramide abundance at the erythrocyte surface was determined utilizing an anti-ceramide antibody. As illustrated in Figure 5, exposure of erythrocytes to 7 U/mL tyrosinase significantly increased the abundance of ceramide at the erythrocyte surface.
The present study uncovers that tyrosinase stimulates cell membrane scrambling leading to phosphatidylserine translocation to the erythrocyte surface. Treatment of human erythrocytes with 5 U/L tyrosinase is further followed by increase of cytosolic Ca2+ activity ([Ca2+]i) and ceramide formation.
Figure 4. Effect of Ca2+ withdrawal on tyrosinase- induced annexin-V-binding. (A,B) Original histograms of annexin V binding erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence 7 U/mL tyrosinase in the presence (A) and absence (B) of calcium. M1 indicates the gating of annexin V binding cells; (C) arithmetic means ± SEM (n = 4) of the percentage of annexin-V-binding erythrocytes after a 24 h treatment with Ringer solution without (white bar) or with (black bars) 7 U/mL tyrosinase in the presence (left bars, +Ca) and absence (right bars, −Ca) of calcium. *** (p < 0.001) indicates significant difference from the absence of tyrosinase (ANOVA) # (p < 0.05) indicates significant difference from the respective values in the presence of Ca2+.
Figure 4. Effect of Ca2+ withdrawal on tyrosinase- induced annexin-V-binding. (A,B) Original histograms of annexin V binding erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence 7 U/mL tyrosinase in the presence (A) and absence (B) of calcium. M1 indicates the gating of annexin V binding cells; (C) arithmetic means ± SEM (n = 4) of the percentage of annexin-V-binding erythrocytes after a 24 h treatment with Ringer solution without (white bar) or with (black bars) 7 U/mL tyrosinase in the presence (left bars, +Ca) and absence (right bars, −Ca) of calcium. *** (p < 0.001) indicates significant difference from the absence of tyrosinase (ANOVA) # (p < 0.05) indicates significant difference from the respective values in the presence of Ca2+.
Toxins 06 01096 g004
Despite its effect on [Ca2+]i, tyrosinase did not decrease but tended to increase the erythrocyte forward scatter. The increase of [Ca2+]i were expected to activate Ca2+ sensitive K+ channels [12] with subsequent K+ exit, cell membrane hyperpolarization, Cl exit and, thus, cellular loss of KCl with osmotically obliged water [13]. Possibly, tyrosinase inhibits the Ca2+ sensitive K+ channels, blocks the Cl channels or stimulates some other mechanism increasing cell volume. Notably, the ionomycin induced erythrocyte shrinkage was not abrogated in the presence of tyrosinase. Excessive erythrocyte swelling may eventually result in rupture of the cell membrane leading to release of cellular hemoglobin, which is filtered in renal glomerula and subsequently occludes renal tubules [75].
Eryptosis is followed by removal of the defective erythrocytes, Phosphatidylserine at the surface of eryptotic cells binds to the respective receptors of phagocytosing cells leading to subsequent engulfment of the affected erythrocytes [12]. Accordingly, eryptotic cells are rapidly cleared from circulating blood [12]. If the accelerated loss of erythrocytes during stimulated eryptosis is not compensated by enhanced formation of new erythrocytes, the clearance of eryptotic erythrocytes from circulating blood results in anemia [12].
Figure 5. Effect of tyrosinase on ceramide formation. (A) Original histogram of ceramide surface abundance of erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence of 7 U/mL tyrosinase; (B) arithmetic means ± SEM (n = 4) of ceramide abundance after a 24-h incubation in Ringer solution without (white bar) or with 7 U/mL tyrosinase (black bar). * (p < 0.05) indicates significant difference from the absence of tyrosinase (t test).
Figure 5. Effect of tyrosinase on ceramide formation. (A) Original histogram of ceramide surface abundance of erythrocytes following exposure for 24 h to Ringer solution without (grey shadow) and with (black line) presence of 7 U/mL tyrosinase; (B) arithmetic means ± SEM (n = 4) of ceramide abundance after a 24-h incubation in Ringer solution without (white bar) or with 7 U/mL tyrosinase (black bar). * (p < 0.05) indicates significant difference from the absence of tyrosinase (t test).
Toxins 06 01096 g005
Phosphatidylserine exposing erythrocytes may further adhere to the vascular wall by binding of phosphatidylserine at the erythrocyte surface to endothelial CXCL16/SR-PSO [76]. The adherence of suicidal erythrocytes to the vascular wall is expected to interfere with microcirculation [76,77,78,79,80,81]. Phosphatidylserine exposing erythrocytes have further been shown to trigger blood clotting and, thus, foster thrombosis [77,82,83].

3. Experimental Section

3.1. Erythrocytes, Solutions and Chemicals

Leukocyte-depleted erythrocytes were kindly provided by the blood bank of the University of Tübingen. The study is approved by the ethics committee of the University of Tübingen (184/2003V). Erythrocytes were incubated in vitro at a hematocrit of 0.4% in Ringer solution containing (in mM) 125 NaCl, 5 KCl, 1 MgSO4, 32 N-2-hydroxyethylpiperazine-N-2-ethanesulfonic acid (HEPES), 5 glucose, 1 CaCl2; pH 7.4 at 37 °C for 48 h. Where indicated, erythrocytes were exposed to mushroom tyrosinase (Sigma, Aldrich, Germany) at the indicated concentrations. In Ca2+-free Ringer solution, 1 mM CaCl2 was substituted by 1 mM glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA).

3.2. FACS Analysis of Annexin-V-Binding and Forward Scatter

After incubation under the respective experimental condition, 50 µL cell suspension was washed in Ringer solution containing 5 mM CaCl2 and then stained with Annexin-V-FITC (1:200 dilution; ImmunoTools, Friesoythe, Germany) in this solution at 37 °C for 20 min under protection from light. In the following, the forward scatter (FSC) of the cells was determined, and annexin-V fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur (BD, Heidelberg, Germany). Following treatment with Ca2+-free Ringer solution, care was taken to measure annexin V binding rapidly enough to avoid triggering of cell membrane by the addition of 5 mM Ca2+.

3.3. Measurement of Intracellular Ca2+

After incubation erythrocytes were washed in Ringer solution and then loaded with Fluo-3/AM (Biotium, Hayward, USA) in Ringer solution containing 5 mM CaCl2 and 5 µM Fluo-3/AM. The cells were incubated at 37 °C for 30 min and washed twice in Ringer solution containing 5 mM CaCl2. The Fluo-3/AM-loaded erythrocytes were resuspended in 200 µL Ringer. Then, Ca2+-dependent fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur.

3.4. Determination of Ceramide Formation

For the determination of ceramide, a monoclonal antibody-based assay was used. After incubation, cells were stained for 1 h at 37 °C with 1 µg/mL anti ceramide antibody (clone MID 15B4, Alexis, Grünberg, Germany) in PBS containing 0.1% bovine serum albumin (BSA) at a dilution of 1:5. The samples were washed twice with PBS-BSA. Subsequently, the cells were stained for 30 min with polyclonal fluorescein isothiocyanate (FITC) conjugated goat anti-mouse IgG and IgM specific antibody (Pharmingen, Hamburg, Germany) diluted 1:50 in PBS-BSA. Unbound secondary antibody was removed by repeated washing with PBS-BSA. The samples were then analyzed by flow cytometric analysis with an excitation wavelength of 488 nm and an emission wavelength of 530 nm.

3.5. Statistics

Data are expressed as arithmetic means ± SEM. As indicated in the figure legends, statistical analysis was made using ANOVA with Tukey’s test as post-test and t-test as appropriate. n denotes the number of different erythrocyte specimens studied. As different erythrocyte specimens used in distinct experiments are differently susceptible to triggers of eryptosis, the same erythrocyte specimens have been used for control and experimental conditions.

4. Conclusions

Tyrosinase stimulates Ca2+ entry, which in turn triggers cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface.

Acknowledgments

The authors acknowledge the meticulous preparation of the manuscript by Ali Soleimanpour and Tanja Loch. The study was supported by the Deutsche Forschungsgemeinschaft and the Open Access Publishing Fund of Tuebingen University.

Author Contributions

Leonie Frauenfeld, Kousi Alzoubi, Majed Abed: have done the experiments, collected the data, prepared the figures. Florian Lang: conception and writing of the paper. All authors have read and approved the final version of the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Seo, S.Y.; Sharma, V.K.; Sharma, N. Mushroom tyrosinase: Recent prospects. J. Agric. Food Chem. 2003, 51, 2837–2853. [Google Scholar] [CrossRef]
  2. Vogel, F.S.; Kemper, L.A.; Jeffs, P.W.; Cass, M.W.; Graham, D.G. Gamma-l-glutaminyl-4-hydroxybenzene, an inducer of cryptobiosis in agaricus bisporus and a source of specific metabolic inhibitors for melanogenic cells. Cancer Res. 1977, 37, 1133–1136. [Google Scholar]
  3. Wick, M.M.; Fitzgerald, G. Inhibition of reverse transcriptase by tyrosinase generated quinones related to levodopa and dopamine. Chem. Biol. Interact. 1981, 38, 99–107. [Google Scholar] [CrossRef]
  4. Wick, M.M.; Rosowsky, A.; Ratliff, J. Antitumor effects of L-glutamic acid dihydroxyanilides against experimental melanoma. J. Invest. Dermatol. 1980, 74, 112–114. [Google Scholar] [CrossRef]
  5. Walton, K.; Walker, R.; Ioannides, C. Effect of baking and freeze-drying on the direct and indirect mutagenicity of extracts from the edible mushroom agaricus bisporus. Food Chem. Toxicol. 1998, 36, 315–320. [Google Scholar] [CrossRef]
  6. Toth, B. Mushroom toxins and cancer (review). Int. J. Oncol. 1995, 6, 137–145. [Google Scholar]
  7. Walton, K.; Coombs, M.M.; Walker, R.; Ioannides, C. Bioactivation of mushroom hydrazines to mutagenic products by mammalian and fungal enzymes. Mutat. Res. 1997, 381, 131–139. [Google Scholar] [CrossRef]
  8. Walton, K.; Coombs, M.M.; Walker, R.; Ioannides, C. The metabolism and bioactivation of agaritine and of other mushroom hydrazines by whole mushroom homogenate and by mushroom tyrosinase. Toxicology 2001, 161, 165–177. [Google Scholar] [CrossRef]
  9. Papaparaskeva, C.; Ioannides, C.; Walker, R. Agaritine does not mediate the mutagenicity of the edible mushroom agaricus bisporus. Mutagenesis 1991, 6, 213–217. [Google Scholar] [CrossRef]
  10. Papaparaskeva-Petrides, C.; Ioannides, C.; Walker, R. Contribution of phenolic and quinonoid structures in the mutagenicity of the edible mushroom agaricus bisporus. Food Chem. Toxicol. 1993, 31, 561–567. [Google Scholar] [CrossRef]
  11. Russo, G.L.; De Nisco, E.; Fiore, G.; Di Donato, P.; d’Ischia, M.; Palumbo, A. Toxicity of melanin-free ink of sepia officinalis to transformed cell lines: Identification of the active factor as tyrosinase. Biochem. Biophys. Res. Commun. 2003, 308, 293–299. [Google Scholar] [CrossRef]
  12. Lang, E.; Qadri, S.M.; Lang, F. Killing me softly—Suicidal erythrocyte death. Int. J. Biochem. Cell Biol. 2012, 44, 1236–1243. [Google Scholar] [CrossRef]
  13. Lang, P.A.; Kaiser, S.; Myssina, S.; Wieder, T.; Lang, F.; Huber, S.M. Role of Ca2+-activated K+ channels in human erythrocyte apoptosis. Am. J. Physiol. Cell Physiol. 2003, 285, C1553–C1560. [Google Scholar] [CrossRef]
  14. Bhavsar, S.K.; Bobbala, D.; Xuan, N.T.; Foller, M.; Lang, F. Stimulation of suicidal erythrocyte death by alpha-lipoic acid. Cell Physiol. Biochem. 2010, 26, 859–868. [Google Scholar] [CrossRef]
  15. Foller, M.; Huber, S.M.; Lang, F. Erythrocyte programmed cell death. IUBMB Life 2008, 60, 661–668. [Google Scholar] [CrossRef]
  16. Foller, M.; Mahmud, H.; Gu, S.; Wang, K.; Floride, E.; Kucherenko, Y.; Luik, S.; Laufer, S.; Lang, F. Participation of leukotriene C(4) in the regulation of suicidal erythrocyte death. J. Physiol. Pharmacol. 2009, 60, 135–143. [Google Scholar]
  17. Lau, I.P.; Chen, H.; Wang, J.; Ong, H.C.; Leung, K.C.; Ho, H.P.; Kong, S.K. In vitro effect of ctab- and peg-coated gold nanorods on the induction of eryptosis/erythroptosis in human erythrocytes. Nanotoxicology 2012, 6, 847–856. [Google Scholar] [CrossRef]
  18. Maellaro, E.; Leoncini, S.; Moretti, D.; Del Bello, B.; Tanganelli, I.; De Felice, C.; Ciccoli, L. Erythrocyte caspase-3 activation and oxidative imbalance in erythrocytes and in plasma of type 2 diabetic patients. Acta Diabetol. 2013, 50, 489–495. [Google Scholar] [CrossRef]
  19. Foller, M.; Sopjani, M.; Koka, S.; Gu, S.; Mahmud, H.; Wang, K.; Floride, E.; Schleicher, E.; Schulz, E.; Munzel, T.; et al. Regulation of erythrocyte survival by amp-activated protein kinase. FASEB J. 2009, 23, 1072–1080. [Google Scholar] [CrossRef]
  20. Kucherenko, Y.; Zelenak, C.; Eberhard, M.; Qadri, S.M.; Lang, F. Effect of casein kinase 1alpha activator pyrvinium pamoate on erythrocyte ion channels. Cell Physiol. Biochem. 2012, 30, 407–417. [Google Scholar] [CrossRef]
  21. Zelenak, C.; Eberhard, M.; Jilani, K.; Qadri, S.M.; Macek, B.; Lang, F. Protein kinase ck1alpha regulates erythrocyte survival. Cell Physiol. Biochem. 2012, 29, 171–180. [Google Scholar] [CrossRef]
  22. Foller, M.; Feil, S.; Ghoreschi, K.; Koka, S.; Gerling, A.; Thunemann, M.; Hofmann, F.; Schuler, B.; Vogel, J.; Pichler, B.; et al. Anemia and splenomegaly in cgki-deficient mice. Proc. Natl. Acad. Sci. USA 2008, 105, 6771–6776. [Google Scholar] [CrossRef] [Green Version]
  23. Bhavsar, S.K.; Gu, S.; Bobbala, D.; Lang, F. Janus kinase 3 is expressed in erythrocytes, phosphorylated upon energy depletion and involved in the regulation of suicidal erythrocyte death. Cell Physiol. Biochem. 2011, 27, 547–556. [Google Scholar] [CrossRef]
  24. Klarl, B.A.; Lang, P.A.; Kempe, D.S.; Niemoeller, O.M.; Akel, A.; Sobiesiak, M.; Eisele, K.; Podolski, M.; Huber, S.M.; Wieder, T.; et al. Protein kinase c mediates erythrocyte “programmed cell death” following glucose depletion. Am. J. Physiol. Cell Physiol. 2006, 290, C244–C253. [Google Scholar]
  25. Gatidis, S.; Zelenak, C.; Fajol, A.; Lang, E.; Jilani, K.; Michael, D.; Qadri, S.M.; Lang, F. P38 MAPK activation and function following osmotic shock of erythrocytes. Cell Physiol. Biochem. 2011, 28, 1279–1286. [Google Scholar] [CrossRef]
  26. Zelenak, C.; Foller, M.; Velic, A.; Krug, K.; Qadri, S.M.; Viollet, B.; Lang, F.; Macek, B. Proteome analysis of erythrocytes lacking amp-activated protein kinase reveals a role of pak2 kinase in eryptosis. J. Proteome Res. 2011, 10, 1690–1697. [Google Scholar] [CrossRef]
  27. Lupescu, A.; Shaik, N.; Jilani, K.; Zelenak, C.; Lang, E.; Pasham, V.; Zbidah, M.; Plate, A.; Bitzer, M.; Foller, M.; et al. Enhanced erythrocyte membrane exposure of phosphatidylserine following sorafenib treatment: An in vivo and in vitro study. Cell Physiol. Biochem. 2012, 30, 876–888. [Google Scholar] [CrossRef]
  28. Shaik, N.; Lupescu, A.; Lang, F. Sunitinib-sensitive suicidal erythrocyte death. Cell Physiol. Biochem. 2012, 30, 512–522. [Google Scholar] [CrossRef]
  29. Abed, M.; Towhid, S.T.; Mia, S.; Pakladok, T.; Alesutan, I.; Borst, O.; Gawaz, M.; Gulbins, E.; Lang, F. Sphingomyelinase-induced adhesion of eryptotic erythrocytes to endothelial cells. Am. J. Physiol. Cell Physiol. 2012, 303, C991–C999. [Google Scholar] [CrossRef]
  30. Abed, M.; Towhid, S.T.; Shaik, N.; Lang, F. Stimulation of suicidal death of erythrocytes by rifampicin. Toxicology 2012, 302, 123–128. [Google Scholar] [CrossRef]
  31. Bottger, E.; Multhoff, G.; Kun, J.F.; Esen, M. Plasmodium falciparum-infected erythrocytes induce granzyme b by nk cells through expression of host-hsp70. PLoS One 2012, 7, e33774. [Google Scholar] [CrossRef]
  32. Firat, U.; Kaya, S.; Cim, A.; Buyukbayram, H.; Gokalp, O.; Dal, M.S.; Tamer, M.N. Increased caspase-3 immunoreactivity of erythrocytes in stz diabetic rats. Exp. Diabetes Res. 2012, 2012, 316384. [Google Scholar]
  33. Ganesan, S.; Chaurasiya, N.D.; Sahu, R.; Walker, L.A.; Tekwani, B.L. Understanding the mechanisms for metabolism-linked hemolytic toxicity of primaquine against glucose 6-phosphate dehydrogenase deficient human erythrocytes: Evaluation of eryptotic pathway. Toxicology 2012, 294, 54–60. [Google Scholar] [CrossRef]
  34. Gao, M.; Cheung, K.L.; Lau, I.P.; Yu, W.S.; Fung, K.P.; Yu, B.; Loo, J.F.; Kong, S.K. Polyphyllin D induces apoptosis in human erythrocytes through Ca2+ rise and membrane permeabilization. Arch. Toxicol. 2012, 86, 741–752. [Google Scholar] [CrossRef]
  35. Ghashghaeinia, M.; Cluitmans, J.C.; Akel, A.; Dreischer, P.; Toulany, M.; Koberle, M.; Skabytska, Y.; Saki, M.; Biedermann, T.; Duszenko, M.; et al. The impact of erythrocyte age on eryptosis. Br. J. Haematol. 2012, 157, 606–614. [Google Scholar] [CrossRef]
  36. Jilani, K.; Lupescu, A.; Zbidah, M.; Abed, M.; Shaik, N.; Lang, F. Enhanced apoptotic death of erythrocytes induced by the mycotoxin ochratoxin A. Kidney Blood Press. Res. 2012, 36, 107–118. [Google Scholar] [CrossRef]
  37. Jilani, K.; Lupescu, A.; Zbidah, M.; Shaik, N.; Lang, F. Withaferin a-stimulated Ca2+ entry, ceramide formation and suicidal death of erythrocytes. Toxicol. in Vitro 2013, 27, 52–58. [Google Scholar] [CrossRef]
  38. Kucherenko, Y.V.; Lang, F. Inhibitory effect of furosemide on non-selective voltage-independent cation channels in human erythrocytes. Cell Physiol. Biochem. 2012, 30, 863–875. [Google Scholar] [CrossRef]
  39. Lang, E.; Qadri, S.M.; Jilani, K.; Zelenak, C.; Lupescu, A.; Schleicher, E.; Lang, F. Carbon monoxide-sensitive apoptotic death of erythrocytes. Basic Clin. Pharmacol. Toxicol. 2012, 111, 348–355. [Google Scholar]
  40. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, E.; Lang, F. Enhanced Ca(2+) entry, ceramide formation, and apoptotic death of erythrocytes triggered by plumbagin. J. Nat. Prod. 2012, 75, 1956–1961. [Google Scholar] [CrossRef]
  41. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, F. Induction of apoptotic erythrocyte death by rotenone. Toxicology 2012, 300, 132–137. [Google Scholar] [CrossRef]
  42. Lupescu, A.; Jilani, K.; Zelenak, C.; Zbidah, M.; Qadri, S.M.; Lang, F. Hexavalent chromium-induced erythrocyte membrane phospholipid asymmetry. Biometals 2012, 25, 309–318. [Google Scholar] [CrossRef]
  43. Polak-Jonkisz, D.; Purzyc, L. Ca influx versus efflux during eryptosis in uremic erythrocytes. Blood Purif. 2012, 34, 209–210. [Google Scholar] [CrossRef]
  44. Qian, E.W.; Ge, D.T.; Kong, S.K. Salidroside protects human erythrocytes against hydrogen peroxide-induced apoptosis. J. Nat. Prod. 2012, 75, 531–537. [Google Scholar] [CrossRef]
  45. Shaik, N.; Zbidah, M.; Lang, F. Inhibition of Ca(2+) entry and suicidal erythrocyte death by naringin. Cell Physiol. Biochem. 2012, 30, 678–686. [Google Scholar] [CrossRef]
  46. Vota, D.M.; Maltaneri, R.E.; Wenker, S.D.; Nesse, A.B.; Vittori, D.C. Differential erythropoietin action upon cells induced to eryptosis by different agents. Cell Biochem. Biophys. 2013, 65, 145–157. [Google Scholar] [CrossRef]
  47. Weiss, E.; Cytlak, U.M.; Rees, D.C.; Osei, A.; Gibson, J.S. Deoxygenation-induced and Ca(2+) dependent phosphatidylserine externalisation in red blood cells from normal individuals and sickle cell patients. Cell Calcium 2012, 51, 51–56. [Google Scholar] [CrossRef]
  48. Zappulla, D. Environmental stress, erythrocyte dysfunctions, inflammation, and the metabolic syndrome: Adaptations to CO2 increases? J. Cardiometab Syndr. 2008, 3, 30–34. [Google Scholar] [CrossRef]
  49. Zbidah, M.; Lupescu, A.; Jilani, K.; Lang, F. Stimulation of suicidal erythrocyte death by fumagillin. Basic Clin. Pharmacol. Toxicol. 2013, 112, 346–351. [Google Scholar] [CrossRef]
  50. Zbidah, M.; Lupescu, A.; Shaik, N.; Lang, F. Gossypol-induced suicidal erythrocyte death. Toxicology 2012, 302, 101–105. [Google Scholar] [CrossRef]
  51. Zelenak, C.; Pasham, V.; Jilani, K.; Tripodi, P.M.; Rosaclerio, L.; Pathare, G.; Lupescu, A.; Faggio, C.; Qadri, S.M.; Lang, F. Tanshinone IIA stimulates erythrocyte phosphatidylserine exposure. Cell Physiol. Biochem. 2012, 30, 282–294. [Google Scholar] [CrossRef]
  52. Abed, M.; Herrmann, T.; Alzoubi, K.; Pakladok, T.; Lang, F. Tannic acid induced suicidal erythrocyte death. Cell Physiol. Biochem. 2013, 32, 1106–1116. [Google Scholar] [CrossRef]
  53. Ahmed, M.S.; Langer, H.; Abed, M.; Voelkl, J.; Lang, F. The uremic toxin acrolein promotes suicidal erythrocyte death. Kidney Blood Press. Res. 2013, 37, 158–167. [Google Scholar] [CrossRef]
  54. Ghashghaeinia, M.; Cluitmans, J.C.; Toulany, M.; Saki, M.; Koberle, M.; Lang, E.; Dreischer, P.; Biedermann, T.; Duszenko, M.; Lang, F.; et al. Age sensitivity of NFκB abundance and programmed cell death in erythrocytes induced by NFκB inhibitors. Cell Physiol. Biochem. 2013, 32, 801–813. [Google Scholar] [CrossRef]
  55. Abed, M.; Feger, M.; Alzoubi, K.; Pakladok, T.; Frauenfeld, L.; Geiger, C.; Towhid, S.T.; Lang, F. Sensitization of erythrocytes to suicidal erythrocyte death following water deprivation. Kidney Blood Press. Res. 2013, 37, 567–578. [Google Scholar]
  56. Alzoubi, K.; Honisch, S.; Abed, M.; Lang, F. Triggering of suicidal erythrocyte death by penta-o-galloyl-beta-d-glucose. Toxins 2014, 6, 54–65. [Google Scholar] [CrossRef]
  57. Jilani, K.; Qadri, S.M.; Lang, F. Geldanamycin-induced phosphatidylserine translocation in the erythrocyte membrane. Cell Physiol. Biochem. 2013, 32, 1600–1609. [Google Scholar]
  58. Jilani, K.; Lang, F. Carmustine-induced phosphatidylserine translocation in the erythrocyte membrane. Toxins 2013, 5, 703–716. [Google Scholar] [CrossRef]
  59. Jilani, K.; Enkel, S.; Bissinger, R.; Almilaji, A.; Abed, M.; Lang, F. Fluoxetine induced suicidal erythrocyte death. Toxins 2013, 5, 1230–1243. [Google Scholar] [CrossRef]
  60. Bissinger, R.; Modicano, P.; Frauenfeld, L.; Lang, E.; Jacobi, J.; Faggio, C.; Lang, F. Estramustine-induced suicidal erythrocyte death. Cell Physiol. Biochem. 2013, 32, 1426–1436. [Google Scholar] [CrossRef]
  61. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, F. Patulin-induced suicidal erythrocyte death. Cell Physiol. Biochem. 2013, 32, 291–299. [Google Scholar] [CrossRef]
  62. Lupescu, A.; Bissinger, R.; Jilani, K.; Lang, F. Triggering of suicidal erythrocyte death by celecoxib. Toxins 2013, 5, 1543–1554. [Google Scholar] [CrossRef]
  63. Lang, E.; Modicano, P.; Arnold, M.; Bissinger, R.; Faggio, C.; Abed, M.; Lang, F. Effect of thioridazine on erythrocytes. Toxins 2013, 5, 1918–1931. [Google Scholar] [CrossRef]
  64. Calderon-Salinas, J.V.; Munoz-Reyes, E.G.; Guerrero-Romero, J.F.; Rodriguez-Moran, M.; Bracho-Riquelme, R.L.; Carrera-Gracia, M.A.; Quintanar-Escorza, M.A. Eryptosis and oxidative damage in type 2 diabetic mellitus patients with chronic kidney disease. Mol. Cell Biochem. 2011, 357, 171–179. [Google Scholar] [CrossRef]
  65. Nicolay, J.P.; Schneider, J.; Niemoeller, O.M.; Artunc, F.; Portero-Otin, M.; Haik, G., Jr.; Thornalley, P.J.; Schleicher, E.; Wieder, T.; Lang, F. Stimulation of suicidal erythrocyte death by methylglyoxal. Cell Physiol. Biochem. 2006, 18, 223–232. [Google Scholar] [CrossRef]
  66. Myssina, S.; Huber, S.M.; Birka, C.; Lang, P.A.; Lang, K.S.; Friedrich, B.; Risler, T.; Wieder, T.; Lang, F. Inhibition of erythrocyte cation channels by erythropoietin. J. Am. Soc. Nephrol. 2003, 14, 2750–2757. [Google Scholar] [CrossRef]
  67. Lang, P.A.; Beringer, O.; Nicolay, J.P.; Amon, O.; Kempe, D.S.; Hermle, T.; Attanasio, P.; Akel, A.; Schafer, R.; Friedrich, B.; et al. Suicidal death of erythrocytes in recurrent hemolytic uremic syndrome. J. Mol. Med. (Berl.) 2006, 84, 378–388. [Google Scholar] [CrossRef]
  68. Kempe, D.S.; Akel, A.; Lang, P.A.; Hermle, T.; Biswas, R.; Muresanu, J.; Friedrich, B.; Dreischer, P.; Wolz, C.; Schumacher, U.; et al. Suicidal erythrocyte death in sepsis. J. Mol. Med. 2007, 85, 269–277. [Google Scholar]
  69. Foller, M.; Bobbala, D.; Koka, S.; Huber, S.M.; Gulbins, E.; Lang, F. Suicide for survival–death of infected erythrocytes as a host mechanism to survive malaria. Cell Physiol. Biochem. 2009, 24, 133–140. [Google Scholar] [CrossRef]
  70. Lang, P.A.; Kasinathan, R.S.; Brand, V.B.; Duranton, C.; Lang, C.; Koka, S.; Shumilina, E.; Kempe, D.S.; Tanneur, V.; Akel, A.; et al. Accelerated clearance of plasmodium-infected erythrocytes in sickle cell trait and annexin-A7 deficiency. Cell Physiol. Biochem. 2009, 24, 415–428. [Google Scholar] [CrossRef]
  71. Lang, P.A.; Schenck, M.; Nicolay, J.P.; Becker, J.U.; Kempe, D.S.; Lupescu, A.; Koka, S.; Eisele, K.; Klarl, B.A.; Rubben, H.; et al. Liver cell death and anemia in wilson disease involve acid sphingomyelinase and ceramide. Nat. Med. 2007, 13, 164–170. [Google Scholar] [CrossRef]
  72. Kempe, D.S.; Lang, P.A.; Duranton, C.; Akel, A.; Lang, K.S.; Huber, S.M.; Wieder, T.; Lang, F. Enhanced programmed cell death of iron-deficient erythrocytes. FASEB J. 2006, 20, 368–370. [Google Scholar]
  73. Qadri, S.M.; Mahmud, H.; Lang, E.; Gu, S.; Bobbala, D.; Zelenak, C.; Jilani, K.; Siegfried, A.; Foller, M.; Lang, F. Enhanced suicidal erythrocyte death in mice carrying a loss-of-function mutation of the adenomatous polyposis coli gene. J. Cell Mol. Med. 2012, 16, 1085–1093. [Google Scholar] [CrossRef]
  74. Birka, C.; Lang, P.A.; Kempe, D.S.; Hoefling, L.; Tanneur, V.; Duranton, C.; Nammi, S.; Henke, G.; Myssina, S.; Krikov, M.; et al. Enhanced susceptibility to erythrocyte “apoptosis” following phosphate depletion. Pflugers Arch. 2004, 448, 471–477. [Google Scholar]
  75. Harrison, H.E.; Bunting, H.; Ordway, N.K.; Albrink, W.S. The pathogenesis of the renal injury produced in the dog by hemoglobin or methemoglobin. J. Exp. Med. 1947, 86, 339–356. [Google Scholar] [CrossRef]
  76. Borst, O.; Abed, M.; Alesutan, I.; Towhid, S.T.; Qadri, S.M.; Foller, M.; Gawaz, M.; Lang, F. Dynamic adhesion of eryptotic erythrocytes to endothelial cells via CXCL16/SR-PSOX. Am. J. Physiol. Cell Physiol. 2012, 302, C644–C651. [Google Scholar] [CrossRef]
  77. Andrews, D.A.; Low, P.S. Role of red blood cells in thrombosis. Curr. Opin. Hematol. 1999, 6, 76–82. [Google Scholar] [CrossRef]
  78. Closse, C.; Dachary-Prigent, J.; Boisseau, M.R. Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br. J. Haematol. 1999, 107, 300–302. [Google Scholar] [CrossRef]
  79. Gallagher, P.G.; Chang, S.H.; Rettig, M.P.; Neely, J.E.; Hillery, C.A.; Smith, B.D.; Low, P.S. Altered erythrocyte endothelial adherence and membrane phospholipid asymmetry in hereditary hydrocytosis. Blood 2003, 101, 4625–4627. [Google Scholar] [CrossRef]
  80. Pandolfi, A.; Di Pietro, N.; Sirolli, V.; Giardinelli, A.; Di Silvestre, S.; Amoroso, L.; Di Tomo, P.; Capani, F.; Consoli, A.; Bonomini, M. Mechanisms of uremic erythrocyte-induced adhesion of human monocytes to cultured endothelial cells. J. Cell Physiol. 2007, 213, 699–709. [Google Scholar] [CrossRef]
  81. Wood, B.L.; Gibson, D.F.; Tait, J.F. Increased erythrocyte phosphatidylserine exposure in sickle cell disease: Flow-cytometric measurement and clinical associations. Blood 1996, 88, 1873–1880. [Google Scholar]
  82. Chung, S.M.; Bae, O.N.; Lim, K.M.; Noh, J.Y.; Lee, M.Y.; Jung, Y.S.; Chung, J.H. Lysophosphatidic acid induces thrombogenic activity through phosphatidylserine exposure and procoagulant microvesicle generation in human erythrocytes. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 414–421. [Google Scholar]
  83. Zwaal, R.F.; Comfurius, P.; Bevers, E.M. Surface exposure of phosphatidylserine in pathological cells. Cell Mol. Life Sci. 2005, 62, 971–988. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Frauenfeld, L.; Alzoubi, K.; Abed, M.; Lang, F. Stimulation of Erythrocyte Cell Membrane Scrambling by Mushroom Tyrosinase. Toxins 2014, 6, 1096-1108. https://doi.org/10.3390/toxins6031096

AMA Style

Frauenfeld L, Alzoubi K, Abed M, Lang F. Stimulation of Erythrocyte Cell Membrane Scrambling by Mushroom Tyrosinase. Toxins. 2014; 6(3):1096-1108. https://doi.org/10.3390/toxins6031096

Chicago/Turabian Style

Frauenfeld, Leonie, Kousi Alzoubi, Majed Abed, and Florian Lang. 2014. "Stimulation of Erythrocyte Cell Membrane Scrambling by Mushroom Tyrosinase" Toxins 6, no. 3: 1096-1108. https://doi.org/10.3390/toxins6031096

Article Metrics

Back to TopTop